You are on page 1of 14

The n e w e ng l a n d j o u r na l of m e dic i n e

Review Article

Dan L. Longo, M.D., Editor

Rheumatoid Arthritis — Common Origins,


Divergent Mechanisms
Ellen M. Gravallese, M.D., and Gary S. Firestein, M.D.​​

R
heumatoid arthritis is one of the most common immune-medi- From the Division of Rheumatology, In-
ated diseases. Its primary manifestation is inflammatory arthritis charac- flammation, and Immunity, Brigham and
Women’s Hospital, Boston (E.M.G.); and
terized by symmetric, polyarticular pain and swelling, typically involving the Division of Rheumatology, Allergy,
the small joints of the hands and feet. However, rheumatoid arthritis is a sys- and Immunology, University of California
temic disease associated with multiple coexisting conditions and extraarticular at San Diego School of Medicine, La Jolla
(G.S.F.). Dr. Gravallese can be contacted
manifestations. Onset of inflammatory synovitis results from the interactions of at ­egravallese@​­bwh​.­harvard​.­edu.
genetic factors and specific environmental exposures. The disease process begins
N Engl J Med 2023;388:529-42.
years before clinically apparent arthritis and manifests as a continuum that origi- DOI: 10.1056/NEJMra2103726
nates with asymptomatic immune dysfunction and progresses through various Copyright © 2023 Massachusetts Medical Society.
stages before the disease can be classified as rheumatoid arthritis.
This review focuses on seropositive rheumatoid arthritis, marked by the pres- CME
at NEJM.org
ence of autoantibodies to post-translationally modified proteins, including anti–
citrullinated protein antibodies (ACPAs, measured as anti–cyclic citrullinated
peptide antibodies); less specific autoantibodies, known as rheumatoid factors,
that bind the Fc portion of immunoglobulin; or both antibody types. Seronegative
rheumatoid arthritis is a separate entity marked by polyarthritis but with poorly
defined pathogenetic mechanisms. The course of seronegative rheumatoid arthri-
tis is typically less destructive to joints,1 but the approach to treatment is similar
to that of seropositive disease.
In contrast to an immune disease such as psoriasis, which largely depends on
the dominant interleukin-23–interleukin-17 pathway, rheumatoid arthritis has
multiple potential paths to a common clinical presentation. The disease progress-
es from preclinical rheumatoid arthritis through chronic disease and involves
pathogenic pathways and cell lineages that can differ among patients, complicat-
ing therapeutic efforts. The predominance of certain pathways over others in in-
dividual patients is underscored by the diversity of clinical responses to targeted
therapies, despite a remarkably similar clinical phenotype. There have been revolu-
tionary changes in the treatment of rheumatoid arthritis in the past three decades,
but many patients still have persistent disease. The ability to identify the specific
pathogenic mechanisms in individual patients would improve outcomes by direct-
ing therapy to those targets.
The preclinical stages of seropositive rheumatoid arthritis are characterized by
disordered immunity, often associated with mucosal surfaces, including the oral
cavity, lungs, and gastrointestinal tract, and by local and systemic generation of
ACPAs. These autoantibodies can be detected in the blood a median of 4.5 years
before the onset of arthritis.2 The risk of rheumatoid arthritis increases over time
as autoantibody levels increase. As this preclinical phase progresses, ACPAs directed
against an expanding array of protein epitopes ensue, along with a rise in pro­
inflammatory proteins in blood, ultimately resulting in joint inflammation.3
Immune responses to altered peptides are not limited to citrullination; carbamy­la­
tion, malon­dialdehyde–acetaldehyde adduct formation, and other protein modifi-

n engl j med 388;6  nejm.org  February 9, 2023 529


The New England Journal of Medicine
Downloaded from nejm.org at FIOCRUZ on February 9, 2023. For personal use only. No other uses without permission.
Copyright © 2023 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Risk factors
• Genetics
• Environmental
exposures Potential
(including smoking) treatment Cell lineage or preclinical
for disease driver-specific mechanisms
• Mucosal
inflammation prevention • Synovial inflammation
• Protein modification • Antigen-driven T- and
(e.g., citrullination) • Second hit B-cell immunity
• Autoantibody • Multiple possible • Fibroblast activation Window of opportunity for
production mechanisms • Epigenetic remodeling treatment to prevent chronicity

Preclinical RA RA Early RA Established


Healthy RA
At risk for RA Transition Clinical synovitis
Personalized mechanisms:
• Immune activation • T cells • New adaptive T-cell Joint damage
and loss of • B cells subsets
Osteoclasts
tolerance • Fibroblasts • Synovial and proteases
• Increased • Inflammatory cytokines autoantibody
autoantibody levels production RA flare
• Other cell types
and affinity • New fibroblast Preinflammatory
• Epigenetic subpopulations and mesenchymal
remodeling aggressive behavior (PRIME) cells

Figure 1. Initiation and Progression of Rheumatoid Arthritis (RA).


The boxes show various stages of the disease continuum and the pathogenic features of each stage. The purple text shows characteristics
that affect the transition between disease stages. RA progresses from a healthy state to preclinical RA (at risk for RA) to the RA transition
to early synovitis and finally to established, destructive disease. The pathway is not unidirectional, since persons in the disease stage
before synovitis who are positive for antibodies against citrullinated peptides (ACPAs) can become ACPA-negative, and in some ACPA-
positive persons, disease never develops. The continuum of disease evolution offers potential opportunities for the prevention of RA.
Although each disease state has a characteristic clinical phenotype, multiple pathways and mechanisms can contribute to pathogenesis
for an individual patient. This is depicted by the red text and arrows, which indicate disease that is dependent on a particular cell type
or mediator. Therapeutic approaches should ideally be targeted to address the particular pathogenic mechanism in an individual patient.
Some patients may have disease that is characterized by multiple mechanisms, resulting in a partial response or a lack of response to a
given targeted therapy.

cations can induce recognition of neoantigens, 1.0%, although the prevalence is higher in cer-
with the production of antibodies to these modi-
tain populations, such as Indigenous North
fied protein antigens.4 Americans. Rheumatoid arthritis can occur at
Treatments are designed to induce clinical any age, but the incidence peaks in the third
remission in patients with established rheuma- through fifth decades of life, and the disease is
toid arthritis. In addition, disease prevention2 to 3 times as common among women as it is
strategies are being developed for persons con-among men. The effects of estrogen on immune
sidered to be at risk for the disease on the basis
function probably play a part in the female pre-
of family history, autoantibody status, geneticdominance of the disease,5 although additional
risk factors, or a combination of these find- sex-related factors are also likely to be involved.
ings, as well as for persons with very early Several infectious agents have been proposed
stages of joint pain or inflammation, before as etiologic or contributing agents, including
rheumatoid arthritis has been definitively diag-
Epstein–Barr virus, retroviruses, bacterial super-
nosed (Fig. 1). antigens, and mycoplasma species, as well as
organisms such as oral Porphyromonas gingivalis
and gut prevotella species.6,7 However, a single
Epidemiol o gy a nd Dise a se
Cl a ssific at ion microorganism that accounts for all patients is
unlikely to be causal. The most prominent be-
The prevalence of rheumatoid arthritis is re- havioral risk factor for the development of rheu-
markably consistent worldwide, at about 0.5 to matoid arthritis is cigarette smoking. Additional

530 n engl j med 388;6 nejm.org February 9, 2023

The New England Journal of Medicine


Downloaded from nejm.org at FIOCRUZ on February 9, 2023. For personal use only. No other uses without permission.
Copyright © 2023 Massachusetts Medical Society. All rights reserved.
Rheumatoid Arthritis

factors marginally increase the risk of rheuma- More than 100 additional alleles have been
toid arthritis, including obesity, low vitamin D identified that contribute to the risk of disease
levels, and use of oral contraceptives. Factors and overwhelmingly implicate immune pathways.
that decrease the risk include a Mediterranean Many are located in gene regulatory or intronic
diet, n–3 fatty acid intake, fish oil supplementa- regions, but some involve the coding region and
tion, and alcohol consumption.8,9 affect gene function. For example, a polymor-
Although rheumatoid arthritis is characteris- phism in PTPN22, a phosphatase involved in T-cell
tically marked by symmetric arthritis in the receptor signaling, is one of the best-character-
small joints of the hands and feet, as the disease ized alleles associated with rheumatoid arthritis.
progresses, any synovial joint can be involved. R620W, a gain-of-function amino acid change in
The 2010 American College of Rheumatology– PTPN22, increases disease risk by a factor of
European League against Rheumatism classifi- more than 2.15 Many other risk alleles are also
cation criteria10 focus on earlier disease manifes- associated with immune processes, including the
tations than did previous classification criteria, coding region of the interleukin-6 receptor and
with the introduction of a composite scoring noncoding regions near the TRAF1–C5 locus.
system that includes the number and site of Most of these alleles marginally increase the
clinically involved joints, the duration of symp- odds ratio for rheumatoid arthritis, by a factor
toms, and the status with respect to rheumatoid of approximately 1.1 to 1.2.
factor, ACPAs, and acute-phase reactants. ACPAs The relatively low concordance of rheumatoid
are increasingly used to support the diagnosis arthritis in monozygotic twins (approximately
because of their high specificity. 15%), as compared with the concordance of mono-
genic diseases, suggests that noncoding DNA
epigenetic marks, possibly induced by environ-
Gene t ic R isk a nd Epigene t ic
Fac t or s mental or stochastic factors, are also important.
DNA methylation might contribute to disease sus-
The most prominent risk factor for rheumatoid ceptibility, as suggested by distinct methylation
arthritis is genetic. For first-degree relatives of patterns in twins who are discordant for rheu-
patients with rheumatoid arthritis, the risk of matoid arthritis.16 Furthermore, in at-risk persons
disease is increased by a factor of 2 to 5. The without synovitis who have high blood levels of
HLA-DR locus is the most important genetic rheumatoid factor or ACPAs, peripheral-blood
association. Well-characterized sequences in the mononuclear cells are characterized by abnormal
hypervariable region of the HLA-DRβ chain DNA methylation in immune-related genes years
(amino acids 70–74), known as the “shared epi- before the onset of symptoms.17 Later, T cells with
tope,” are associated with an increased risk. aberrant epigenetic marks in immunologic path-
HLA-DR is involved in antigen presentation to ways accumulate in the inflamed synovium.18 In
CD4+ T cells and could increase susceptibility contrast, patients with osteoarthritis have fewer
through its ability to bind and present specific differentially marked genes in synovial T cells,
arthritogenic peptides. HLA-DR genes associated and they are randomly distributed. Thus, remodel-
with rheumatoid arthritis can bind peptides mod- ing of the disease-associated epigenome in
ified by citrullination more avidly than native synovium could be driven by processes that con-
peptides, inducing T-cell activation and cytokine tribute to the transition from preclinical to
production. In addition, these HLA molecules clinical rheumatoid arthritis.
may influence T-cell receptor selection toward a
more autoimmune repertoire.11 Informatics analy- From Muc os a l Infl a m m at ion
sis of major histocompatibility complex (MHC) t o A lter ed Pep t ide s t o Cl inic a l
data indicates that three amino acid positions Dise a se
in HLA-DRβ1 and a single amino acid in HLA-B
and HLA-DPβ1 that modify the peptide-binding Environmental and behavioral influences play a
groove explain most of the MHC association major role in susceptibility to rheumatoid arthri-
with disease risk.12 HLA-DRB1 is associated not tis and disease severity. Cigarette smoking and
only with susceptibility but also with disease genetic risk can be synergistic: for ACPA-positive
severity and possibly with varying treatment smokers with two copies of the susceptibility
responses to certain biologic agents.13,14 shared epitope, the risk of rheumatoid arthritis

n engl j med 388;6  nejm.org  February 9, 2023 531


The New England Journal of Medicine
Downloaded from nejm.org at FIOCRUZ on February 9, 2023. For personal use only. No other uses without permission.
Copyright © 2023 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

is 20 times that for nonsmokers.19 The risk gradu- coincide with synovitis has not been identified
ally abates after smoking cessation, approaching in patients with rheumatoid arthritis. Synovial-
the risk for nonsmokers within two to three biopsy specimens from persons with preclinical
decades.20 Inflammation and stress at mucosal rheumatoid arthritis and arthralgias show little
surfaces, induced by environmental exposures or no evidence of inflammation or local immune
such as cigarette smoke, contribute to disease responses despite high levels of circulating
initiation in persons with risk alleles for rheu- ACPAs.28 In preclinical models, the administra-
matoid arthritis, and the link between mucosal tion of ACPA autoantibodies does not cause ar-
inflammation and rheumatoid arthritis is stron- thritis29 but can exacerbate existing synovitis.
gest for the airway. The mammalian genome in- Even so, rising ACPA titers in humans are a
cludes enzymes known as peptidyl arginine harbinger of clinical disease. When the titers in
deiminases, which convert arginine to citrulline. at-risk persons reach 3 times the upper limit of
These enzymes are induced by cell stress and the normal range, there is a 30 to 50% chance
lead to post-translational citrullination of many that rheumatoid arthritis will be diagnosed
proteins. Citrullination is quite active in the air- within 3 to 5 years.30
ways of smokers, where modified peptides have These observations raise the possibility that
been detected in macrophages.21 progression from preclinical rheumatoid arthri-
Bronchiolar thickening and local neutrophil tis to established disease might be prevented
extracellular trap formation occur in asymptom- through therapeutic intervention or mitigation
atic at-risk persons with high ACPA titers, as well of environmental stress. Several clinical trials un-
as in first-degree relatives of patients with rheu- successfully attempted to intercede in this tran-
matoid arthritis.22 The extruded DNA in neutro- sition, including treatment with atorvastatin31
phil extracellular traps forms a scaffold for citrul- and B-cell depletion with a single course of
linated peptides and amplifies immune responses rituximab.32 The latter delayed, but did not pre-
that can generate ACPAs.23 In at-risk persons, the vent, conversion to clinical rheumatoid arthritis
combination of peptide citrullination and HLA- in ACPA-positive persons presenting with arthral-
DR haplotypes that bind citrullinated peptides gias. In at-risk persons with arthralgias and
more avidly than native peptides24 can lead to a imaging evidence of synovitis, 1 year of treat-
local immune response and further ACPA produc- ment with methotrexate, a nonadaptive immune-
tion. However, ACPAs produced at sites of muco- system intervention, did not prevent rheumatoid
sal damage might also serve as a mechanism to arthritis, as assessed after 2 years. However, the
clear citrullinated proteins.25 The evolving ACPAs disease was less severe in the treated cohort.33
arise from B cells and plasmablasts through af- Although proinflammatory processes have been
finity maturation, which is driven by specific emphasized in the transition to clinical disease,
citrullinated proteins and oligoclonal expansion of inadequate production of antiinflammatory cyto-
antigen-specific cells.26 Concomitant increases kines, such as interleukin-1 receptor antagonist
in serum cytokines and chemokines provide evi- (interleukin-1Ra) and interleukin-10, or defective
dence of early systemic inflammation that ulti- synovial apoptosis34 could also contribute to the
mately culminates in symptomatic joint inflam- onset and perpetuation of disease.
mation.
Production of ACPAs and other autoantibod- He tero genei t y of S y nov i t is
ies represents a break in tolerance. Such breaks in R heum at oid A r thr i t is
can be facilitated by the selective introduction of
N-linked glycosylation sites in the B-cell recep- Synovitis is a hallmark of rheumatoid arthritis,
tor antigen-binding pocket, which alters the anti- with an influx of inflammatory cells leading to
gen-binding site and enhances B-cell activation.27 multiple villous projections within the joint cav-
ACPAs (IgA or IgG) can bind to an array of ity. Typical histologic features include synovial
citrullinated proteins, including fibronectin, hyperplasia, neovascularization, and a heteroge-
enolase, histones, and fibrinogen. The mere neous inflammatory infiltrate that can include
presence of ACPAs is not sufficient to induce lymphoid aggregates and germinal center–like
arthritis, and a consistent pattern of citrullinat- structures. Infiltrating cells include T and B cells,
ed proteins or antibody levels that precede or plasma cells, plasmablasts, macrophages, den-

532 n engl j med 388;6  nejm.org  February 9, 2023

The New England Journal of Medicine


Downloaded from nejm.org at FIOCRUZ on February 9, 2023. For personal use only. No other uses without permission.
Copyright © 2023 Massachusetts Medical Society. All rights reserved.
Rheumatoid Arthritis

dritic cells, and occasional mast cells and natu- Ne w Insigh t s in t o Patho gene sis
ral killer cells. Neutrophils are sparse in rheuma-
toid synovium but pass through tissues into Studies of specimens from ultrasound-guided
synovial fluid rapidly. synovial biopsy in patients with rheumatoid ar-
Synovial histologic and transcriptional analy- thritis offer new insights into pathogenesis.
ses show marked heterogeneity among patients Evaluation of synovial cell surface markers with
with established rheumatoid arthritis,35 perhaps cytometry by time-of-flight and single-cell RNA
providing clues to pathogenic pathways that are sequencing has provided data on the large array
active in a given patient. Synovial assessments, of cell lineages in rheumatoid synovium, includ-
however, can be complicated by sampling bias ing more than 20 transcription-defined T-cell
and by distinct epigenetic marks and transcrip- subtypes.42 T cells are central in the pathogene-
tomes that depend on joint location.36 For exam- sis of rheumatoid arthritis, and one important
ple, fibroblasts derived from hip and knee syno- remaining question is whether some of these
via can be distinguished from each other on the cell phenotypes are responsible for the disease,
basis of DNA methylation and transcriptome represent a response to the synovial microenvi-
patterns. Noncoding RNAs also vary according ronment, or are merely “spectators at a fire,”
to joint location and could help shape stromal recruited by the rich chemoattractant milieu.
cell phenotypes and function.37 Although classifi- Several novel and important T-cell subsets have
cation systems have been proposed on the basis been identified, however, including peripheral
of histologic features or the most prominent cell helper T (Tph) cells, located within synovial B-cell
types in a given patient’s synovial tissue on bi- clusters and in the circulation, which promote
opsy,38 meaningful correlations between histo- B-cell production of interleukin-21, supporting
pathological patterns and clinical disease activ- immunoglobulin affinity maturation, among
ity or outcomes are thus far limited. Tissue other functions (Table 1). Tph cells also promote
analyses performed with RNA sequencing meth- B-cell proliferation and differentiation into anti-
ods and stratification by cell lineage signatures body-producing plasma cells.43 Oligoclonal ex-
may have the potential for predicting the response pansion of synovial B cells with somatic muta-
to a given therapy.39 Data from synovial biopsies tions, indicating local ACPA affinity maturation,
in patients with rheumatoid arthritis suggest is also prominent in rheumatoid synovium.26
that molecular and histologic profiling might Studies using fate mapping systems have
provide insights into the response to the B-cell– identified novel macrophage subsets, including
directed agent rituximab as compared with block- CX3CR1+ tissue-resident macrophages that form
ade of the interleukin-6 receptor with tocilizu­ an immunologic barrier on the synovial surface,
mab.40 These approaches continue to be promising restricting the flux of proteins across the normal
research tools but have not yet defined specific synovial lining.48 Identification of additional
pathways driving disease in a given patient. macrophage subsets, including resident macro-
Approaches based on systems biology might phages47 with an antiinflammatory phenotype
also help stratify patients according to shared and inflammatory macrophages that contribute
pathogenic pathways. A recent study integrated to the production of proinflammatory factors,
transcription factor–binding site accessibility also provides insights into pathogenesis. In ad-
with the transcriptome in fibroblasts.41 At least dition, dendritic cells, which are present in rheu-
two clusters of patients were identified on the matoid synovium, play a part in local antigen pre-
basis of divergent transcription factor functions sentation and activation of autoreactive T cells.56
in cultured fibroblast-like synoviocytes. For ex- Extensive work with synovial biopsy specimens
ample, the transcription factor retinoic acid re- from patients with rheumatoid arthritis has
ceptor alpha had proproliferative effects in the highlighted the importance of several novel fibro-
transforming growth factor β pathway in one blast phenotypes that promote inflammation,
cluster but antiproliferative effects in the other including those with proinflammatory functions
cluster. Similar unbiased systems approaches whose differentiation is regulated by endothelial
could provide insight into how biologic features cells.51 In addition, the transcription factor ETS1
vary among patients with similar clinical pheno- defines a fibroblast phenotype that regulates
types. bone damage through the production of RANKL

n engl j med 388;6  nejm.org  February 9, 2023 533


The New England Journal of Medicine
Downloaded from nejm.org at FIOCRUZ on February 9, 2023. For personal use only. No other uses without permission.
Copyright © 2023 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Table 1. Recently Identified Cell Lineages and Cell Phenotypes Contributing to Rheumatoid Arthritis (RA).*

Cell Class Comments Study


T cells
CD4+ PD1+ CXCR5− (Tph) Located in lymphoid aggregates adjacent to B cells; produce interleukin-21, Rao et al.43
which supports B-cell proliferation and differentiation into plasma cells
CD8+ GZMK+ Located in RA synovial sublining; source of interferon-γ Jonsson et al.44
B cells
Mucosal, circulating, and Oligoclonal expansion with evidence of recirculation and ACPA affinity matura- Kongpachith et al.26
synovial B cells tion due to somatic mutation
NR4A+ Produces lymphotoxins and interleukin-6, which promote lymphoid aggregate Meednu et al.45
formation
Macrophages
MerTK− Proinflammatory; found in active RA synovium; associated with interleukin-6 Alivernini et al.46
and TNF production
MerTK+ Antiinflammatory; associated with lipoxin and resolvin production Cai et al.47
CX3CR1+ Resident cells in RA synovial lining with tight junctions that serve as immuno- Culemann et al.48
logic barrier; disrupted in RA synovium
Alternatively activated Interleukin-33 reprogrammed with metabolic rewiring that uncouples respira- Faas et al.49
tory chain and enhances inflammation resolution
HBEGF+ Promotes fibroblast aggressiveness and invasion Kuo et al.50
Fibroblasts
FAP+ CD90+ Proinflammatory phenotype located in sublining; regulated by NOTCH3 Wei et al.51
FAP+ CD90− Located in intimal lining; produce interleukin-6, metalloproteinases, and pros- Mizoguchi et al.52
tanoids in RA
ETS1 Regulates bone damage through production of RANKL by fibroblasts in synovium Yan et al.53
PRIME cells Present in circulation of patients with RA (according to transcriptome profile); Orange et al.54
increase in peripheral-blood PRIME cells precedes RA flares, possibly asso­
ciated with B-cell activation
Neutrophils: synovium and lung Produce NETs, which bind citrullinated peptides to enhance ACPA production Corsiero et al.55
mucosa

* ACPA denotes anti–citrullinated protein antibody, NETs neutrophil extracellular traps, PRIME preinflammatory mesenchymal, RANKL recep-
tor activator of nuclear factor-κB ligand, TNF tumor necrosis factor, and Tph peripheral helper T.

(receptor activator of nuclear factor-κB ligand) L ong -Ter m C onsequence s


and activation of osteoclasts.53 of Dise a se
Murine studies indicate that fibroblasts have
the potential to migrate from an inflamed joint Morbidity and Mortality
through the bloodstream and could thus “spread” Patients with rheumatoid arthritis have an in-
synovitis.57 More recent studies in humans have creased risk of death. Cardiovascular disease is
identified the appearance of preinflammatory the most common cause of premature death,
mesenchymal (PRIME) cells in peripheral blood and the excess risk of cardiovascular disease in
obtained just before disease flares in patients rheumatoid arthritis is attributed to the combi-
with rheumatoid arthritis.54 Longitudinal tran- nation of chronic inflammation and well-docu-
scriptomic data also provide evidence of periph- mented risk factors for cardiovascular disease
eral-blood B-cell activation before the appear- such as hypertension and dyslipidemia. Tradi-
ance of PRIME cells and disease flares. tional risk factors (e.g., elevated low-density lipo-
Interactions between B cells and PRIME cells protein cholesterol levels) have a weaker correla-
could therefore serve as a potential target to tion with cardiovascular risk in rheumatoid
abrogate disease flares. arthritis than in the general population.58 Recent

534 n engl j med 388;6  nejm.org  February 9, 2023

The New England Journal of Medicine


Downloaded from nejm.org at FIOCRUZ on February 9, 2023. For personal use only. No other uses without permission.
Copyright © 2023 Massachusetts Medical Society. All rights reserved.
Rheumatoid Arthritis

Figure 2. Complications of RA and Coexisting Conditions.


Proinflammatory cytokines, complement activation, Stroke
and immune complex formation in RA promote sys- Depression, mental
temic inflammation that affects many systems and re- health disorders
sults in complications and coexisting conditions, along
with the production of acute-phase reactants (APRs) Scleritis, episcleritis
mediated in large part by interleukin-6. Designation as Sicca syndrome
a complication or coexisting condition is not exact,
and the two categories can overlap. Altered lipid me- Bronchial inflammation,
tabolism and cytokines, including tumor necrosis fac- bronchiectasis
tor and interleukin-6, contribute to atherogenesis, Interstitial lung disease
myocardial infarction, and stroke. Pulmonary compli-
cations occur through multiple mechanisms. Rheuma- Myocardial infarction
toid nodules and vasculitis are seen less commonly Congestive heart failure
Valvular abnormalities
with current treatment approaches. Chronic inflamma-
Pericarditis
tion contributes to metabolic and other coexisting con-
ditions and can result in depression and altered coping
behaviors. Possible mechanisms affecting the immune Lymphoproliferative
disease
status of the central nervous system (CNS) include
effects of proinflammatory cytokines on activation of
Lipid abnormalities,
blood–brain barrier endothelium, leading to the trans- APRs
port of cytokines into the CNS, and effects on neural
circuits and plasticity.62 Proinflammatory cytokines
Vasculitis
synergize with RANKL (receptor activator of nuclear Nodules
factor-κB ligand) to promote osteoclastogenesis and
articular and systemic bone loss, leading to fractures,
Obesity
and inhibitors of the Wnt signaling pathway prevent
bone formation and erosion repair by osteoblasts. Joint
destruction is mitigated by tight control of inflamma- Joint damage
tion. The inflamed synovium produces multiple algo-
gens that increase pain sensitivity by reducing the fir- Low bone mineral
ing threshold for local nociceptors. Sensitization of density, osteoporosis,
fracture
central pain pathways by proinflammatory mediators
has also been implicated.63 The majority of patients
Pain
with RA have marked fatigue due to pain, sleep distur-
Functional deterioration
bance, and other factors.64 These complications and
Infection
coexisting conditions combine to result in a multifac-
torial deterioration of function over time. Susceptibility
to infections is increased in RA owing to impaired host
defense, and this can be exacerbated by the use of im- and progress in some patients, and are associ-
munosuppressive agents. ated with a higher risk of death from respiratory
disease.65 A variant in the promoter region of the
gene encoding mucin 5B (MUC5B) is associated
studies have shown coronary microvascular dys- with idiopathic pulmonary fibrosis, and this vari-
function in patients with rheumatoid arthritis, ant is also associated with the usual interstitial
like that seen in patients with diabetes,59 which pneumonia form of interstitial lung disease in
probably contributes to excess cardiovascular patients with rheumatoid arthritis,66 implicating
mortality. Current treatment strategies that re- mucins in the pathogenesis of this complication.
duce inflammation mitigate the risk of death.
The benefit of this approach is particularly well Joint Destruction
documented with tumor necrosis factor (TNF) The rheumatoid synovium is characterized by
blockers.60 expansion of tissue at the interface with carti-
Complications and coexisting conditions in lage and bone. This expanding tissue, known as
patients with rheumatoid arthritis, including an pannus, resembles a locally invasive tumor and
increased risk of lymphoproliferative disease,61 extends over the surface of cartilage. Pannus
are shown in Figure 2. As rheumatoid arthritis also invades the bone marrow space directly or
progresses, pulmonary interstitial fibrosis, bron- through pores in cortical bone. In active rheu-
chial inflammation, and bronchiectasis develop matoid arthritis, the extracellular matrix of

n engl j med 388;6 nejm.org February 9, 2023 535


The New England Journal of Medicine
Downloaded from nejm.org at FIOCRUZ on February 9, 2023. For personal use only. No other uses without permission.
Copyright © 2023 Massachusetts Medical Society. All rights reserved.
536
Table 2. Agents Approved and in General Use or Not Approved for Use in RA.*

Class and Agent Mechanism of Action Comments


Approved and in general use
Traditional DMARDs
Methotrexate Affects multiple cell types; inhibits several pathways, including adenosine Used as a single agent or in combination ther­
­metabolism apy; most common initial agent
The

Leflunomide Inhibits dihydroorotate dehydrogenase and pyrimidine metabolism and may Used as a single agent or in combination
inhibit expansion of activated leukocytes therapy
Sulfasalazine Combination drug (5-aminosalicylic acid and sulfapyridine) that potentially inhibits Often used in combination with methotrexate
inflammatory cytokines and chemokines and alters adenosine metabolism and hydroxychloroquine
Hydroxychloroquine Possibly stabilizes macrophage lysosomes; modulates TLR7 and TLR9 activity Used as monotherapy for mild disease; often
used in combination with methotrexate
and sulfasalazine (triple therapy)
Biologic DMARDs
Cytokine inhibition Interruption of cytokine networks Often used in combination with methotrexate
or another traditional DMARD
TNF: adalimumab, certolizumab pegol, Blockade of TNF inhibits activation of leukocytes, FLS, endothelial cells, and os-
n e w e ng l a n d j o u r na l

etanercept, golimumab, infliximab teoclasts, preventing matrix degradation and production of proinflammatory

The New England Journal of Medicine


of

molecules
Interleukin-6: sarilumab, tocilizumab Blockade inhibits B-cell differentiation; activation of leukocytes, osteoclasts, and
acute-phase reactant elevation; lipid alterations

n engl j med 388;6  nejm.org  February 9, 2023


Interleukin-1: anakinra Interleukin-1Ra blocks interleukin-1 binding to receptor; inhibits activation of leuko- Less effective than other anticytokine agents

Copyright © 2023 Massachusetts Medical Society. All rights reserved.


cytes, FLS, endothelial cells, and osteoclasts, preventing matrix degradation in RA
m e dic i n e

T cells: abatacept Binds CD80 and CD86 and blocks T-cell costimulation, inhibiting naive T-cell Increased efficacy when used in combination
­activation with methotrexate
B cells: rituximab Binds CD20 and depletes B cells, inhibiting antigen presentation and autoantibody Often used in combination with methotrexate
production
Synthetic DMARDs–JAK inhibitors: baricitinib, Interrupt cytokine networks through blockade of JAK–STAT pathway, inhibiting FLS
tofacitinib, upadacitinib activation, leukocyte maturation, and autoantibody production

Downloaded from nejm.org at FIOCRUZ on February 9, 2023. For personal use only. No other uses without permission.
Selected biologic DMARD targets tested but
agent not approved
Cytokine inhibition Interruption of cytokine networks
Class and Agent Mechanism of Action Comments
GM-CSF Blockade inhibits differentiation and activation of myeloid cells and granulocytes Efficacy in phase 2 studies; phase 3 studies in
progress
Interleukin-15 Blockade inhibits T-cell–FLS and T-cell–macrophage interactions and TNF Modest efficacy in clinical trials
­production
Interleukin-17A Blockade inhibits synergism with TNF and activation of FLS, osteoclasts, and Modest efficacy in phase 3 trials
­chondrocytes
Interleukin-18 Blockade inhibits granulocyte and osteoclast activation and Th1 responses Minimal efficacy
Interleukin-23 Blockade inhibits Th17 expansion Modest efficacy in phase 3 studies
Interferon-γ Blockade inhibits cytokine production and class II MHC antigen induction Ineffective
Lymphotoxin alpha Blockade inhibits lymphoid organ architecture Ineffective
RANKL Blockade inhibits osteoclast differentiation and systemic and articular bone loss Effective in phase 2 trials for erosions but not
clinical synovitis
Signal transduction
p38 MAP kinase Blockade interrupts cytokine networks Modest, transient efficacy
Bruton’s tyrosine kinase Blockade inhibits B-cell receptor signaling and B-cell activation Marginal-to-modest efficacy, depending on the
compound
PI3 kinase γ or δ Blockade inhibits cell proliferation, survival, and migration to synovium Mixed efficacy
Syk tyrosine kinase Blockade inhibits activation of T cells, B cells, macrophages, and FLS Modest efficacy
Cell-targeting
Cadherin-11 Depletes FLS Ineffective
Rheumatoid Arthritis

CD4 Depletes CD4+ T cells Ineffective


CD52 Depletes CD4+ T cells and several other immune-cell lineages Ineffective

The New England Journal of Medicine


n engl j med 388;6  nejm.org  February 9, 2023
CD5 Depletes certain T lymphocytes and mantle-zone lymphocytes Ineffective
Cell recruitment
ICAM-1 Blocks ICAM–αLβ2 integrin interactions, inhibiting cell recruitment to synovium Ineffective
Multiple chemokines and chemokine Block cell recruitment to synovium Limited or no efficacy for antichemokine anti­

Copyright © 2023 Massachusetts Medical Society. All rights reserved.


receptors bodies and small-molecule chemokine
­receptor inhibitors

* DMARD denotes disease-modifying antirheumatic drug, FLS fibroblast-like synoviocytes, GM-CSF granulocyte–macrophage colony-stimulating factor, ICAM-1 intercellular adhesion
molecule 1, interleukin-1Ra interleukin-1 receptor antagonist, JAK–STAT Janus kinase–signal transducer and activator of transcription, MAP mitogen-activated protein, MHC major his-
tocompatibility complex, PI3 phosphatidylinositol 3, Th1 type 1 helper T cell, Th17 type 17 helper T cell, and TLR toll-like receptor.

Downloaded from nejm.org at FIOCRUZ on February 9, 2023. For personal use only. No other uses without permission.
537
The n e w e ng l a n d j o u r na l of m e dic i n e

cartilage, ligaments, and tendons is destroyed by of potentially toxic medications such as nonste-
proteinases that are produced by synovial cells, roidal antiinflammatory drugs, glucocorticoids,
especially fibroblasts, and by chondrocytes them- and opioids is also an important focus. The use
selves. The inflammatory cytokine milieu — of composite disease activity measures, often
most notably, interleukin-1β and TNF — directly called “treat to target,” is a critical component
activates these cells to produce matrix metallo- of the treatment strategy.76 Determining the or-
proteinases, including collagenases, stromelysins der in which drugs are used is not as important
and gelatinases, and ADAMTS5, which contrib- as selecting one of the many outcome measures
ute to cartilage and joint destruction.67 for disease activity that can be incorporated into
Bone destruction requires the action of osteo- the clinical workflow and changing or adding
clasts, which differentiate through the combined therapeutic agents as needed to achieve the tar-
actions of the receptor activator of RANKL,68 get of low disease activity or remission.77 As
and proinflammatory cytokines, especially TNF successful new therapeutic agents have been
and interleukin-6.69 The most important source introduced, guidelines for treatment and evi-
of RANKL, promoting bone loss in rheumatoid denced-based approaches have been updated and
arthritis, is synovial fibroblasts,70 but certain are available to guide clinicians.78,79
T‑cell and B-cell subsets also produce RANKL. Despite the advent of new therapies that tar-
Inflamed synovial tissues and pannus bring in- get a wide array of mechanisms, the mainstay
flammatory cytokines and RANKL-expressing for the initial treatment of rheumatoid arthritis
cells to the bone microenvironment, inducing remains low-dose methotrexate, and 25 to 40%
osteoclastogenesis. Osteoclasts attach to bone, of patients have substantial improvement with
forming an acidic environment that leaches min- methotrexate alone.80 Inadequate responses to
eral from bone, and produce enzymes, including methotrexate require the addition of another
cathepsin K, that degrade the bone matrix. In agent, typically a biologic agent or a Janus kinase
addition, inhibitors of the Wnt signaling path- (JAK) inhibitor. Methotrexate improves the clin-
way prevent osteoblast differentiation and bone ical response to many targeted agents when
repair.71,72 RANKL and proinflammatory cyto- used in combination therapy. Combinations of
kines enter the circulation, promoting systemic traditional agents (triple therapy) with metho-
bone loss and osteoporosis and increasing the trexate, sulfasalazine, and hydroxychloroquine
risk of fractures. Systemic bone loss begins in can achieve adequate responses,81 but adherence
the preclinical phase of rheumatoid arthritis, to the regimen may be challenging. Most drug-
since ACPAs can directly promote osteoclasto- specific toxic effects have been well described,
genesis by triggering Fc receptor activation and such as bone marrow suppression and liver en-
cytokine release from macrophages and acti- zyme abnormalities (e.g., with methotrexate and
vating osteoclasts.73 Improved therapies and an leflunomide) or thrombosis and an increase in
aggressive approach to controlling inflamma- cardiovascular events (e.g., with the JAK inhibi-
tion in patients have reduced the severity of ar- tor tofacitinib, as compared with an anti-TNF
ticular and systemic bone loss in patients with agent).82 Effective drugs typically suppress host
rheumatoid arthritis.74 defenses and are associated with low rates of
serious infections (typically ≤1%).83 Glucocorti-
coids are associated with a dose-dependent risk
Ther a peu t ic C onsider at ions
a nd A pproache s of serious infection84 and contribute to fracture
and other complications over time.
Therapeutic approaches and outcomes in pa- Many targeted agents evaluated in clinical
tients with rheumatoid arthritis have improved trials have limited or no efficacy or have not
dramatically over the past three decades with been approved for clinical use (Table 2), but we
the advent of targeted therapies (Table 2 and have learned much from these trials. The site of
Fig. 3). Early diagnosis and intervention with a action of available therapeutic agents in the con-
disease-modifying antirheumatic drug (DMARD) text of pathogenesis is shown in Figure 3. Treat-
remain the cornerstone of treatment to control ment should not simply suppress inflammation
inflammation, prevent joint and organ damage, but must also address the specific pathogenic
and reduce the risk of death.75 Limiting the use pathway (or pathways) in an individual patient’s

538 n engl j med 388;6  nejm.org  February 9, 2023

The New England Journal of Medicine


Downloaded from nejm.org at FIOCRUZ on February 9, 2023. For personal use only. No other uses without permission.
Copyright © 2023 Massachusetts Medical Society. All rights reserved.
Rheumatoid Arthritis

Fc
Immune receptor
complexes
Interleukin-6,
GM-CSF
Cytokines promote
Bone differentiation to
marrow-derived osteoclasts
monocyte or Interleukin-1,
macrophage interleukin-6, Interferon-γ,
interleukin-15, interleukin-17, Interleukin-1,
interleukin-18,TNF TNF TNF
Monocyte
MHC TNF Interleukin-6 or osteoclast
Autoantibodies CD80 or
class II inhibitors inhibitors precursor
including CD86
rheumatoid TCR
factor CD28 Interleukin-6

T cell T cell Interferon-γ, RANK


help interleukin-17, RANKL
TNF
CD40L
Cytokines JAK inhibitors
CD40
Interleukin-6, Baricitinib Fibroblast
Plasma GM-CSF Tofacitinib
B cell Costimulation Upadacitinib
cell
Osteoclast
B–cell Costimulation differentiation
depletion inhibitor
Dendritic GM-CSF and fusion
Rituximab cell
Abatacept
Interferon-α,
interleukin-12, Osteoclast
interleukin-23,
Anti–modified TGFβ
protein Modified
autoantibodies proteins
Proteases

Neutrophil
Bone erosion and
cartilage loss

Figure 3. Synovial Cellular Interactions, Cytokine Networks, and Sites of Action of Current Therapeutic Agents.
Cell–cell interactions within the synovium are critical components of RA pathogenesis. Red boxes show effective therapeutic agents.
Several cell types (dendritic cells, macrophages, and B cells) can present antigens to T cells, including modified (e.g., citrullinated) pro-
teins, to activate these cells and to induce their differentiation. This results in the production of cytokines that, in turn, activate other,
neighboring cells, including monocytes, macrophages, and synovial fibroblasts, to produce additional proinflammatory cytokines and
factors. Neutrophil extracellular traps in the lungs form a scaffold for citrullinated proteins and amplify immune responses that can gener-
ate ACPAs. Activated B cells differentiate into plasma cells that produce ACPAs and other autoantibodies. RANKL is produced by synovial
fibroblasts but also by certain T- and B-cell subsets, inducing the differentiation of monocytes into bone-resorbing osteoclasts. Osteo-
clastic degradation of bone leads to the joint erosions seen in patients with RA, and proteases induced by inflammatory cytokines lead
to cartilage loss and radiographic narrowing of joint spaces. Interleukin-6 inhibitors include tocilizumab and sarilumab. Tumor necrosis
factor (TNF) inhibitors include adalimumab, certolizumab pegol, etanercept, golimumab, and infliximab. Rituximab is an anti-CD20,
B-cell–depleting agent. Abatacept inhibits T-cell costimulation. Janus kinase (JAK) inhibitors include baricitinib, tofacitinib, and upadaci-
tinib. These are positioned between cells producing cytokines including interleukin-6 and inhibit the JAK–STAT (Janus kinase–signal
transducers and activators of transcription) pathway. GM-CSF denotes granulocyte–macrophage colony-stimulating factor, MHC major
histocompatibility complex, TCR T-cell receptor, TGF transforming growth factor.

disease. Methods of stratifying patients to iden- levels in the blood), tissue analyses (e.g., histo-
tify those who are likely to have a response to a pathological patterns or transcriptome assess-
particular treatment, thus improving drug selec- ments), or genetic markers (e.g., single-nucleo-
tion, are being investigated. To date, however, tide polymorphisms) has been shown to improve
no combination of biomarkers (e.g., cytokine decision making in clinical practice. Rational

n engl j med 388;6 nejm.org February 9, 2023 539


The New England Journal of Medicine
Downloaded from nejm.org at FIOCRUZ on February 9, 2023. For personal use only. No other uses without permission.
Copyright © 2023 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

drug selection remains a critical unmet need, seropositive disease in whom anti-TNF agents
and the development of alternative taxonomies and methotrexate were tapered and stopped, the
based on pathogenesis will be essential. cumulative flare rate at 2 years was 61%, and
Disease mechanisms may also vary with the only 15% of patients had a drug-free remission.91
stage of disease. Epigenetic marks of synovial In addition, therapeutic responses may not be
fibroblasts in early rheumatoid arthritis are recaptured when a treatment is reinitiated. Dis-
quite different from those in later stages of the ease recurrence is likely because most current
disease, and the specificities of ACPAs evolve therapeutic agents target downstream inflam-
over time.85 Thus, individualized therapy might matory mediators rather than resetting the im-
require adjustments for mechanisms that vary as mune system or inducing pathways that resolve
the disease progresses. This concept is support- inflammation.
ed by the observation that early disease is more As we increase our understanding of the im-
responsive to therapy than is late disease86 and munologic continuum from a healthy immune
highlights the importance of early control of system to preclinical rheumatoid arthritis to
inflammation. early and chronic disease, new opportunities for
Clinical remission is the goal of therapy but individualized interventions that treat or prevent
is not realized in most patients with rheumatoid disease should emerge. Interceding at the earli-
arthritis. Tapering or even discontinuing thera- est time points to prevent disease will perhaps
pies in patients with complete responses can be be as important as identifying new targets for
achieved in the short term,87 but unfortunately, long-standing rheumatoid arthritis. New classi-
the disease typically recurs. In patients with fication criteria are needed to harmonize data
early rheumatoid arthritis and low disease activ- from clinical trials and observational studies
ity, clinical predictors of disease flare after involving at-risk persons. At the same time,
DMARD discontinuation include measures of analyses of multiple streams of genomic, pro-
the patient’s functioning and measures of bone teomic, metabolomic, and epigenomic data are
erosion on magnetic resonance imaging.88,89 likely to identify new therapeutic targets and
Rates of disease flare may also differ on the enable clinicians to select the agent that will
basis of autoantibody status, as well as the dura- work best in an individual patient.
tion of remission once DMARDs are tapered.90 Disclosure forms provided by the authors are available with
In a study involving patients with well-controlled the full text of this article at NEJM.org.

References
1. Ajeganova S, Huizinga TW. Rheuma- 7. Möller B, Kollert F, Sculean A, Villiger hypervariable region of T cell receptors.
toid arthritis: seronegative and seroposi- PM. Infection triggers in peridontitis and Nat Genet 2022;​54:​393-402.
tive RA: alike but different? Nat Rev Rheu- the gut in rheumatoid arthritis (RA): 12. Raychaudhuri S, Sandor C, Stahl EA,
matol 2015;​11:​8-9. a complex story about association and et al. Five amino acids in three HLA pro-
2. Nielen MM, van Schaardenburg D, causality. Front Immunol 2020;​11:​1108. teins explain most of the association be-
Reesink HW, et al. Specific autoantibod- 8. Klareskog L, Padyukov L, Lorentzen J, tween MHC and seropositive rheumatoid
ies precede the symptoms of rheumatoid Alfredsson L. Mechanisms of disease: arthritis. Nat Genet 2012;​44:​291-6.
arthritis: a study of serial measurements genetic susceptibility and environmental 13. Viatte S, Plant D, Han B, et al. Asso-
in blood donors. Arthritis Rheum 2004;​ triggers in the development of rheuma- ciation of HLA-DRB1 haplotypes with
50:​380-6. toid arthritis. Nat Clin Pract Rheumatol rheumatoid arthritis severity, mortality,
3. Sokolove J, Bromberg R, Deane KD, 2006;​2:​425-33. and treatment response. JAMA 2015;​313:​
et al. Autoantibody epitope spreading in 9. Gan RW, Bemis EA, Demoruelle MK, 1645-56.
the pre-clinical phase predicts progression et al. The association between omega-3 14. Hirose W, Harigai M, Amano K, et al.
to rheumatoid arthritis. PLoS One 2012;​ fatty acid biomarkers and inflammatory Impact of the HLA-DRB1 shared epitope
7(5):​e35296. arthritis in an anti-citrullinated protein on responses to treatment with tofaci-
4. Toes RE, Huizinga TJW. Update on antibody positive population. Rheumatol- tinib or abatacept in patients with rheu-
autoantibodies to modified proteins. Curr ogy (Oxford) 2017;​56:​2229-36. matoid arthritis. Arthritis Res Ther 2021;​
Opin Rheumatol 2015;​27:​262-7. 10. Aletaha D, Neogi T, Silman AJ, et al. 23:​228.
5. Cutolo M, Straub RH. Sex steroids and 2010 Rheumatoid arthritis classification 15. Lee AT, Li W, Liew A, et al. The
autoimmune rheumatic diseases: state of criteria: an American College of Rheuma- PTPN22 R620W polymorphism associates
the art. Nat Rev Rheumatol 2020;​16:​628- tology/European League Against Rheu- with RF positive rheumatoid arthritis in
44. matism collaborative initiative. Arthritis a dose-dependent manner but not with
6. Costenbader KH, Karlson EW. Epstein- Rheum 2010;​62:​2569-81. HLA-SE status. Genes Immun 2005;​ 6:​
Barr virus and rheumatoid arthritis: is 11. Ishigaki K, Lagattuta KA, Luo Y, 129-33.
there a link? Arthritis Res Ther 2006;​8:​ James EA, Buckner JH, Raychaudhuri S. 16. Svendsen AJ, Gervin K, Lyle R, et al.
204. HLA autoimmune risk alleles restrict the Differentially methylated DNA regions in

540 n engl j med 388;6  nejm.org  February 9, 2023

The New England Journal of Medicine


Downloaded from nejm.org at FIOCRUZ on February 9, 2023. For personal use only. No other uses without permission.
Copyright © 2023 Massachusetts Medical Society. All rights reserved.
Rheumatoid Arthritis

monozygotic twin pairs discordant for oping rheumatoid arthritis: implications based biomarker analysis of the phase 4
rheumatoid arthritis: an epigenome-wide for understanding preclinical rheumatoid R4RA randomized trial. Nat Med 2022;​28:​
study. Front Immunol 2016;​7:​510. arthritis. Arthritis Rheumatol 2014;​ 66:​ 1256-68.
17. Shao X, Hudson M, Colmegna I, et al. 513-22. 41. Ainsworth RI, Hammaker D, Nygaard
Rheumatoid arthritis-relevant DNA methyl- 29. Kuhn KA, Kulik L, Tomooka B, et al. G, et al. Systems-biology analysis of rheu-
ation changes identified in ACPA-positive Antibodies against citrullinated proteins matoid arthritis patients reveals key regu-
asymptomatic individuals using methylome enhance tissue injury in experimental lators and enables mechanism-based
capture sequencing. Clin Epigenetics 2019;​ autoimmune arthritis. J Clin Invest 2006;​ stratification. Nat Commun 2022;​ 13:​
11:​110. 116:​961-73. 6221.
18. Ai R, Boyle DL, Wang W, Firestein GS. 30. Deane KD, Holers VM. Rheumatoid 42. Zhang F, Wei K, Slowikowski K, et al.
Distinct DNA methylation patterns of arthritis pathogenesis, prediction, and Defining inflammatory cell states in
rheumatoid arthritis peripheral blood and prevention: an emerging paradigm shift. rheumatoid arthritis joint synovial tissues
synovial tissue T cells. ACR Open Rheu- Arthritis Rheumatol 2021;​73:​181-93. by integrating single-cell transcriptomics
matol 2021;​3:​127-32. 31. van Boheemen L, Turk S, Beers-Tas and mass cytometry. Nat Immunol 2019;​
19. Klareskog L, Malmström V, Lundberg MV, et al. Atorvastatin is unlikely to pre- 20:​928-42.
K, Padyukov L, Alfredsson L. Smoking, vent rheumatoid arthritis in high risk in- 43. Rao DA, Gurish MF, Marshall JL, et al.
citrullination and genetic variability in dividuals: results from the prematurely Pathologically expanded peripheral T help-
the immunopathogenesis of rheumatoid stopped STAtins to Prevent Rheumatoid er cell subset drives B cells in rheumatoid
arthritis. Semin Immunol 2011;​23:​92-8. Arthritis (STAPRA) trial. RMD Open arthritis. Nature 2017;​542:​110-4.
20. Liu X, Tedeschi SK, Barbhaiya M, et al. 2021;​7:​e001591. 44. Jonsson AH, Zhang F, Dunlap G, et al.
Impact and timing of smoking cessation 32. Gerlag DM, Safy M, Maijer KI, et al. Granzyme K+ CD8 T cells form a core
on reducing risk of rheumatoid arthritis Effects of B-cell directed therapy on the population in inflamed human tissue. Sci
among women in the Nurses’ Health preclinical stage of rheumatoid arthritis: Transl Med 2022;​14(649):​eabo0686.
Studies. Arthritis Care Res (Hoboken) the PRAIRI study. Ann Rheum Dis 2019;​ 45. Meednu N, Rangel-Moreno J, Zhang F,
2019;​71:​914-24. 78:​179-85. et al. Dynamic spectrum of ectopic lym-
21. Makrygiannakis D, Hermansson M, 33. Krijbolder DI, Verstappen M, van Dijk phoid B cell activation and hypermutation
Ulfgren AK, et al. Smoking increases pep- BT, et al. Intervention with methotrexate in the RA synovium characterized by
tidylarginine deiminase 2 enzyme expres- in patients with arthralgia at risk of rheu- NR4A nuclear receptor expression. Cell
sion in human lungs and increases citrul- matoid arthritis to reduce the develop- Rep 2022;​39:​110766.
lination in BAL cells. Ann Rheum Dis ment of persistent arthritis and its disease 46. Alivernini S, MacDonald L, Elmes-
2008;​67:​1488-92. burden (TREAT EARLIER): a randomised, mari A, et al. Distinct synovial tissue mac-
22. Demoruelle MK, Harrall KK, Ho L, double-blind, placebo-controlled, proof- rophage subsets regulate inflammation
et al. Anti-citrullinated protein antibod- of-concept trial. Lancet 2022;​400:​283-94. and remission in rheumatoid arthritis.
ies are associated with neutrophil extra- 34. Firestein GS, Yeo M, Zvaifler NJ. Nat Med 2020;​26:​1295-306.
cellular traps in the sputum in relatives of Apoptosis in rheumatoid arthritis 47. Cai B, Kasikara C, Doran AC, Ramak-
rheumatoid arthritis patients. Arthritis synovium. J Clin Invest 1995;​96:​1631-8. rishnan R, Birge RB, Tabas I. MerTK sig-
Rheumatol 2017;​69:​1165-75. 35. Lewis MJ, Barnes MR, Blighe K, et al. naling in macrophages promotes the
23. Corsiero E, Bombardieri M, Carlotti Molecular portraits of early rheumatoid synthesis of inf lammation resolution
E, et al. Single cell cloning and recombi- arthritis identify clinical and treatment mediators by suppressing CaMKII activity.
nant monoclonal antibodies generation response phenotypes. Cell Rep 2019;​28(9):​ Sci Signal 2018;​11:​eaar3721.
from RA synovial B cells reveal frequent 2455-2470.e5. 48. Culemann S, Grüneboom A, Nicolás-
targeting of citrullinated histones of NETs. 36. Ai R, Hammaker D, Boyle DL, et al. Ávila JA, et al. Locally renewing resident
Ann Rheum Dis 2016;​75:​1866-75. Joint-specific DNA methylation and tran- synovial macrophages provide a protec-
24. Hill JA, Southwood S, Sette A, Jevni- scriptome signatures in rheumatoid ar- tive barrier for the joint. Nature 2019;​572:​
kar AM, Bell DA, Cairns E. Cutting edge: thritis identify distinct pathogenic pro- 670-5.
the conversion of arginine to citrulline cesses. Nat Commun 2016;​7:​11849. 49. Faas M, Ipseiz N, Ackermann J, et al.
allows for a high-affinity peptide interac- 37. Frank-Bertoncelj M, Trenkmann M, IL-33-induced metabolic reprogramming
tion with the rheumatoid arthritis-associ- Klein K, et al. Epigenetically-driven ana- controls the differentiation of alterna-
ated HLA-DRB1*0401 MHC class II mol- tomical diversity of synovial fibroblasts tively activated macrophages and the res-
ecule. J Immunol 2003;​171:​538-41. guides joint-specific fibroblast functions. olution of inflammation. Immunity 2021;​
25. Holers VM, Demoruelle MK, Kuhn Nat Commun 2017;​8:​14852. 54(11):​2531-2546.e5.
KA, et al. Rheumatoid arthritis and the 38. Zhang, F, Jonsson, AH, Nathan, A, 50. Kuo D, Ding J, Cohn IS, et al. HBEGF+
mucosal origins hypothesis: protection et al. Cellular deconstruction of inflamed macrophages in rheumatoid arthritis in-
turns to destruction. Nat Rev Rheumatol synovium defines diverse inflammatory duce fibroblast invasiveness. Sci Transl
2018;​14:​542-57. phenotypes in rheumatoid arthritis. Feb- Med 2019;​11(491):​eaau8587.
26. Kongpachith S, Lingampalli N, Ju C-H, ruary 28, 2022 (https://www​.­biorxiv​.­org/​ 51. Wei K, Korsunsky I, Marshall JL, et al.
et al. Affinity maturation of the anti- ­content/​­10​.­1101/​­2022​.­02​.­25​.­481990v1). Notch signalling drives synovial fibro-
citrullinated protein antibody paratope preprint. blast identity and arthritis pathology. Na-
drives epitope spreading and polyreactiv- 39. Humby F, Durez P, Buch MH, et al. ture 2020;​582:​259-64.
ity in rheumatoid arthritis. Arthritis Rituximab versus tocilizumab in anti- 52. Mizoguchi F, Slowikowski K, Wei K,
Rheumatol 2019;​71:​507-17. TNF inadequate responder patients with et al. Functionally distinct disease-associ-
27. Kissel T, Ge C, Hafkenscheid L, et al. rheumatoid arthritis (R4RA): 16-week ated fibroblast subsets in rheumatoid ar-
Surface Ig variable domain glycosylation outcomes of a stratified, biopsy-driven, thritis. Nat Commun 2018;​9:​789.
affects autoantigen binding and acts as multicentre, open-label, phase 4 random­ 53. Yan M, Komatsu N, Muro R, et al.
threshold for human autoreactive B cell ised controlled trial. Lancet 2021;​397:​305- ETS1 governs pathological tissue-remod-
activation. Sci Adv 2022;​8(6):​eabm1759. 17. eling programs in disease-associated fi-
28. de Hair MJ, van de Sande MGH, Ram- 40. Rivellese F, Surace AEA, Goldmann K, broblasts. Nat Immunol 2022;​23:​1330-41.
wadhdoebe TH, et al. Features of the et al. Rituximab versus tocilizumab in 54. Orange DE, Yao V, Sawicka K, et al.
synovium of individuals at risk of devel- rheumatoid arthritis: synovial biopsy- RNA identification of PRIME cells pre-

n engl j med 388;6  nejm.org  February 9, 2023 541


The New England Journal of Medicine
Downloaded from nejm.org at FIOCRUZ on February 9, 2023. For personal use only. No other uses without permission.
Copyright © 2023 Massachusetts Medical Society. All rights reserved.
Rheumatoid Arthritis

dicting rheumatoid arthritis flares. N Engl thritis is a source of osteoclast differen- Therapies for active rheumatoid arthritis
J Med 2020;​383:​218-28. tiation factor. Arthritis Rheum 2000;​43:​ after methotrexate failure. N Engl J Med
55. Corsiero E, Pratesi F, Prediletto E, 250-8. 2013;​369:​307-18.
Bombardieri M, Migliorini P. NETosis as 69. O’Brien W, Fissel BM, Maeda Y, et al. 82. Ytterberg SR, Bhatt DL, Mikuls TR,
source of autoantigens in rheumatoid ar- RANK-independent osteoclast formation et al. Cardiovascular and cancer risk
thritis. Front Immunol 2016;​7:​485. and bone erosion in inflammatory arthri- with tofacitinib in rheumatoid arthritis.
56. Wehr P, Purvis H, Law SC, Thomas R. tis. Arthritis Rheumatol 2016;​ 68:​2889- N Engl J Med 2022;​386:​316-26.
Dendritic cells, T cells and their interac- 900. 83. Lortholary O, Fernandez-Ruiz M,
tion in rheumatoid arthritis. Clin Exp Im- 70. Danks L, Komatsu N, Guerrini MM, Baddley JW, Manuel O, Mariette X, Win-
munol 2019;​196:​12-27. et al. RANKL expressed on synovial fi- throp KL. Infectious complications of
57. Lefèvre S, Knedla A, Tennie C, et al. broblasts is primarily responsible for rheumatoid arthritis and psoriatic arthri-
Synovial fibroblasts spread rheumatoid bone erosions during joint inflammation. tis during targeted and biological thera-
arthritis to unaffected joints. Nat Med Ann Rheum Dis 2016;​75:​1187-95. pies: a viewpoint in 2020. Ann Rheum Dis
2009;​15:​1414-20. 71. Walsh NC, Reinwald S, Manning CA, 2020;​79:​1532-43.
58. Weber B, He Z, Yang N, et al. Diver- et al. Osteoblast function is compromised 84. George MD, Baker JF, Winthrop K,
gence of cardiovascular biomarkers of at sites of focal bone erosion in inflam- et al. Risk for serious infection with low-
lipids and subclinical myocardial injury matory arthritis. J Bone Miner Res 2009;​ dose glucocorticoids in patients with
among rheumatoid arthritis patients with 24:​1572-85. rheumatoid arthritis: a cohort study. Ann
increased inflammation. Arthritis Rheu- 72. Diarra D, Stolina M, Polzer K, et al. Intern Med 2020;​173:​870-8.
matol 2021;​73:​970-9. Dickkopf-1 is a master regulator of joint 85. Ai R, Whitaker JW, Boyle DL, et al.
59. Liao KP, Huang J, He Z, et al. Coro- remodeling. Nat Med 2007;​13:​156-63. DNA methylome signature in synovio-
nary microvascular dysfunction in rheu- 73. Harre U, Georgess D, Bang H, et al. cytes from patients with early rheumatoid
matoid arthritis compared to diabetes Induction of osteoclastogenesis and bone arthritis compared to synoviocytes from
mellitus and association with all-cause loss by human autoantibodies against patients with longstanding rheumatoid
mortality. Arthritis Care Res (Hoboken) citrullinated vimentin. J Clin Invest 2012;​ arthritis. Arthritis Rheumatol 2015;​ 67:​
2021;​73:​159-65. 122:​1791-802. 1978-80.
60. Greenberg JD, Kremer JM, Curtis JR, 74. Lindner L, Callhoff J, Alten R, et al. 86. Emery P, Solem C, Majer I, Cappelleri
et al. Tumour necrosis factor antagonist Osteoporosis in patients with rheumatoid JC, Tarallo M. A European chart review
use and associated risk reduction of car- arthritis: trends in the German National study on early rheumatoid arthritis treat-
diovascular events among patients with Database 2007-2017. Rheumatol Int 2020;​ ment patterns, clinical outcomes, and
rheumatoid arthritis. Ann Rheum Dis 40:​2005-12. healthcare utilization. Rheumatol Int 2015;​
2011;​70:​576-82. 75. Combe B, Landewe R, Daien CI, et al. 35:​1837-49.
61. Baecklund E, Iliadou A, Askling J, et al. 2016 update of the EULAR recommenda- 87. Lillegraven S, Paulshus Sundlisæter N,
Association of chronic inflammation, not tions for the management of early arthri- Aga A-B, et al. Effect of half-dose vs stable-
its treatment, with increased lymphoma tis. Ann Rheum Dis 2017;​76:​948-59. dose conventional synthetic disease-modi-
risk in rheumatoid arthritis. Arthritis 76. Smolen JS, Breedveld FC, Burmester fying antirheumatic drugs on disease flares
Rheum 2006;​54:​692-701. GR, et al. Treating rheumatoid arthritis to in patients with rheumatoid arthritis in
62. Nerurkar L, Siebert S, McInnes IB, Ca- target: 2014 update of the recommenda- remission: the ARCTIC REWIND random­
vanagh J. Rheumatoid arthritis and de- tions of an international task force. Ann ized clinical trial. JAMA 2021;​325:​1755-
pression: an inflammatory perspective. Rheum Dis 2016;​75:​3-15. 64.
Lancet Psychiatry 2019;​6:​164-73. 77. Grigor C, Capell H, Stirling A, et al. 88. Ahmad HA, Baker JF, Conaghan PG,
63. Iyer P, Lee YC. Why it hurts: the mech- Effect of a treatment strategy of tight con- et al. Prediction of flare following remis-
anisms of pain in rheumatoid arthritis. trol for rheumatoid arthritis (the TICORA sion and treatment withdrawal in early
Rheum Dis Clin North Am 2021;​47:​229- study): a single-blind randomised con- rheumatoid arthritis: post hoc analysis of
44. trolled trial. Lancet 2004;​364:​263-9. a phase IIIb trial with abatacept. Arthritis
64. Druce KL, Basu N. Predictors of fa- 78. Fraenkel L, Bathon JM, England BR, Res Ther 2022;​24:​47.
tigue in rheumatoid arthritis. Rheumatol- et al. 2021 American College of Rheuma- 89. Rayner F, Anderson AE, Baker KF,
ogy (Oxford) 2019;​58:​Suppl 5:​v29-v34. tology guideline for the treatment of et al. BIOlogical Factors that Limit sus-
65. Dong H, Julien PJ, Demoruelle MK, rheumatoid arthritis. Arthritis Rheuma- tAined Remission in rhEumatoid arthritis
Deane KD, Weisman MH. Interstitial lung tol 2021;​73:​1108-23. (the BIO-FLARE study): protocol for a
abnormalities in patients with early rheu- 79. Smolen JS, Landewé R, Breedveld FC, non-randomised longitudinal cohort study.
matoid arthritis: a pilot study evaluating et al. EULAR recommendations for the BMC Rheumatol 2021;​5:​22.
prevalence and progression. Eur J Rheu- management of rheumatoid arthritis with 90. Figueiredo CP, Bang H, Cobra JF, et al.
matol 2018;​6:​193-8. synthetic and biological disease-modify- Antimodified protein antibody response
66. Juge PA, Lee JS, Ebstein E, et al. ing antirheumatic drugs: 2013 update. pattern influences the risk for disease re-
MUC5B Promoter variant and rheumatoid Ann Rheum Dis 2014;​73:​492-509. lapse in patients with rheumatoid arthri-
arthritis with interstitial lung disease. 80. Emery P, Bingham CO III, Burmester tis tapering disease modifying antirheu-
N Engl J Med 2018;​379:​2209-19. GR, et al. Certolizumab pegol in combi- matic drugs. Ann Rheum Dis 2017;​ 76:​
67. Gravallese EM, Darling JM, Ladd AL, nation with dose-optimised methotrexate 399-407.
Katz JN, Glimcher LH. In situ hybridiza- in DMARD-naïve patients with early, active 91. van Mulligen E, Weel AE, Hazes JM,
tion studies of stromelysin and collage- rheumatoid arthritis with poor prognos- van der Helm-van Mil A, de Jong PHP. Ta-
nase messenger RNA expression in rheu- tic factors: 1-year results from C-EARLY, a pering towards DMARD-free remission in
matoid synovium. Arthritis Rheum 1991;​ randomised, double-blind, placebo-con- established rheumatoid arthritis: 2-year
34:​1076-84. trolled phase III study. Ann Rheum Dis results of the TARA trial. Ann Rheum Dis
68. Gravallese EM, Manning C, Tsay A, 2017;​76:​96-104. 2020;​79:​1174-81.
et al. Synovial tissue in rheumatoid ar- 81. O’Dell JR, Mikuls TR, Taylor TH, et al. Copyright © 2023 Massachusetts Medical Society.

542 n engl j med 388;6  nejm.org  February 9, 2023

The New England Journal of Medicine


Downloaded from nejm.org at FIOCRUZ on February 9, 2023. For personal use only. No other uses without permission.
Copyright © 2023 Massachusetts Medical Society. All rights reserved.

You might also like